Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Neurophysiol ; 39(4): 276-282, 2022 May 01.
Article in English | MEDLINE | ID: mdl-32804879

ABSTRACT

PURPOSE: Previous work has shown that quantitative EEG measures correlate with the severity of ischemic stroke. This has not been systematically validated in patients with acute ischemic stroke who have undergone mechanical thrombectomy. METHODS: Data were collected from 73 patients who underwent mechanical thrombectomy and had a standard head set EEG performed during their hospital admission. For each patient, the global delta-alpha ratio (DAR) and its difference between the two hemispheres were calculated. Associations between the global and interhemispheric DAR difference with the patients' National Institutes of Health Stroke and Modified Rankin Scale scores at discharge and 3 months after thrombectomy were assessed. RESULTS: The interhemispheric DAR difference correlated with the National Institutes of Health Stroke scores at discharge (Spearman R = 0.41, P = 0.0008), National Institutes of Health Stroke scores at 3 months (Spearman R = 0.60, P = 0.02) and Modified Rankin Scale scores at 3 months (Spearman R = 0.27, P = 0.01). In contrast, the global DAR did not correlate significantly with any of these clinical outcomes when evaluated as continuous variables. In a multivariate logistic regression model, both the interhemispheric DAR difference (ß = 0.25, P = 0.03) and the infarct volume (ß = 0.02, P = 0.03) were independently predictive of good versus poor functional outcome (Modified Rankin Scale score ≤2 vs. >2) at 3 months. CONCLUSIONS: The quantitative EEG measure of interhemispheric slow relative to fast frequencies power asymmetry correlated with the discharge and 3-month National Institutes of Health Stroke and Modified Rankin Scale scores and provided added value to infarct volume in predicting functional outcome at 3 months. These data support the prognostic value of quantitative EEG in ischemic stroke patients who have undergone mechanical thrombectomy.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Brain Ischemia/diagnosis , Brain Ischemia/surgery , Electroencephalography , Humans , Infarction , Prognosis , Retrospective Studies , Stroke/diagnosis , Stroke/surgery , Thrombectomy , Treatment Outcome
2.
Brain Res ; 1657: 185-192, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27916440

ABSTRACT

The stress-hyperresponsive Wistar-Kyoto (WKY) rat strain exhibits a hyperalgesic phenotype and is a useful genetic model for studying stress-pain interactions. Peroxisome proliferator-activated receptor (PPAR) signalling in the midbrain periaqueductal grey (PAG) modulates pain. This study characterised PPAR signalling in the PAG of WKY rats exposed to the formalin test of inflammatory pain, versus Sprague-Dawley (SD) controls. Formalin injection reduced levels of the endogenous PPAR ligands N-palmitoylethanolamide (PEA) and N-oleoylethanolamide (OEA) in the lateral(l) PAG of SD rats, but not WKY rats which exhibited higher levels of these analytes compared with formalin-injected SD counterparts. Levels of mRNA coding for fatty acid amide hydrolase (FAAH; catabolises PEA and OEA) were lower in the lPAG of WKY versus SD rats. PPARγ mRNA and protein levels in the lPAG were higher in saline-treated WKY rats, with PPARγ protein levels reduced by formalin treatment in WKY rats only. In the dorsolateral(dl) or ventrolateral(vl) PAG, there were no effects of formalin injection on PEA or OEA levels but there were some differences in levels of these analytes between saline-treated WKY and SD rats and some formalin-evoked alterations in levels of PPARα, PPARγ or FAAH mRNA in WKY and/or SD rats. Pharmacological blockade of PPARγ in the lPAG enhanced formalin-evoked nociceptive behaviour in WKY, but not SD, rats. These data indicate differences in the PPAR signalling system in the PAG of WKY versus SD rats and suggest that enhanced PEA/OEA-mediated tone at PPARγ in the lPAG may represent an adaptive mechanism to lower hyperalgesia in WKY rats.


Subject(s)
Depression/metabolism , Hyperalgesia/metabolism , Pain Perception/physiology , Periaqueductal Gray/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Stress, Psychological/metabolism , Affect/physiology , Animals , Genetic Predisposition to Disease , Male , Nociceptive Pain/metabolism , Pain Perception/drug effects , Periaqueductal Gray/drug effects , Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors , Rats, Inbred WKY , Rats, Sprague-Dawley , Resilience, Psychological , Signal Transduction , Species Specificity
3.
Pharmacol Res ; 113(Pt A): 44-54, 2016 11.
Article in English | MEDLINE | ID: mdl-27520401

ABSTRACT

Negative affective state has a significant impact on pain, and genetic background is an important moderating influence on this interaction. The Wistar-Kyoto (WKY) inbred rat strain exhibits a stress-hyperresponsive, anxiety/depressive-like phenotype and also displays a hyperalgesic response to noxious stimuli. Transient receptor potential subfamily V member 1 (TRPV1) within the midbrain periaqueductal grey (PAG) plays a key role in regulating both aversive and nociceptive behaviour. In the present study, we investigated the role of TRPV1 in the sub-columns of the PAG in formalin-evoked nociceptive behaviour in WKY versus Sprague-Dawley (SD) rats. TRPV1 mRNA expression was significantly lower in the dorsolateral (DL) PAG and higher in the lateral (L) PAG of WKY rats, compared with SD counterparts. There were no significant differences in TRPV1 mRNA expression in the ventrolateral (VL) PAG between the two strains. TRPV1 mRNA expression significantly decreased in the DLPAG and increased in the VLPAG of SD, but not WKY rats upon intra-plantar formalin administration. Intra-DLPAG administration of either the TRPV1 agonist capsaicin, or the TRPV1 antagonist 5'-Iodoresiniferatoxin (5'-IRTX), significantly increased formalin-evoked nociceptive behaviour in SD rats, but not in WKY rats. The effects of capsaicin were likely due to TRPV1 desensitisation, given their similarity to the effects of 5'-IRTX. Intra-VLPAG administration of capsaicin or 5'-IRTX reduced nociceptive behaviour in a moderate and transient manner in SD rats, and similar effects were seen with 5'-IRTX in WKY rats. Intra-LPAG administration of 5'-IRTX reduced nociceptive behaviour in a moderate and transient manner in SD rats, but not in WKY rats. These results indicate that modulation of inflammatory pain by TRPV1 in the PAG occurs in a sub-column-specific manner. The data also provide evidence for differences in the expression of TRPV1, and differences in the effects of pharmacological modulation of TRPV1 in specific PAG sub-columns, between WKY and SD rats, suggesting that TRPV1 expression and/or functionality in the PAG plays a role in hyper-responsivity to noxious stimuli in a genetic background prone to negative affect.


Subject(s)
Inflammation/metabolism , Pain/metabolism , Periaqueductal Gray/metabolism , TRPV Cation Channels/metabolism , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Behavior, Animal/physiology , Capsaicin/pharmacology , Depression/metabolism , Diterpenes/pharmacology , Genotype , Male , Periaqueductal Gray/drug effects , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY , Rats, Sprague-Dawley
4.
Article in English | MEDLINE | ID: mdl-25988529

ABSTRACT

Repeated exposure to a homotypic stressor such as forced swimming enhances nociceptive responding in rats. However, the influence of genetic background on this stress-induced hyperalgesia is poorly understood. The aim of the present study was to compare the effects of repeated forced swim stress on nociceptive responding in Sprague-Dawley (SD) rats versus the Wistar Kyoto (WKY) rat strain, a genetic background that is susceptible to stress, negative affect and hyperalgesia. Given the well-documented role of the endocannabinoid system in stress and pain, we investigated associated alterations in endocannabinoid signalling in the dorsal horn of the spinal cord and amygdala. In SD rats, repeated forced swim stress for 10 days was associated with enhanced late phase formalin-evoked nociceptive behaviour, compared with naive, non-stressed SD controls. In contrast, WKY rats exposed to 10 days of swim stress displayed reduced late phase formalin-evoked nociceptive behaviour. Swim stress increased levels of monoacylglycerol lipase (MAGL) mRNA in the ipsilateral side of the dorsal spinal cord of SD rats, an effect not observed in WKY rats. In the amygdala, swim stress reduced anandamide (AEA) levels in the contralateral amygdala of SD rats, but not WKY rats. Additional within-strain differences in levels of CB1 receptor and fatty acid amide hydrolase (FAAH) mRNA and levels of 2-arachidonylglycerol (2-AG) were observed between the ipsilateral and contralateral sides of the dorsal horn and/or amygdala. These data indicate that the effects of repeated stress on inflammatory pain-related behaviour are different in two rat strains that differ with respect to stress responsivity and affective state and implicate the endocannabinoid system in the spinal cord and amygdala in these differences.


Subject(s)
Endocannabinoids/metabolism , Nociceptive Pain/physiopathology , Rats, Inbred WKY/physiology , Rats, Sprague-Dawley/physiology , Stress, Psychological/physiopathology , Amygdala/physiopathology , Animals , Disease Models, Animal , Formaldehyde , Functional Laterality , Genetic Predisposition to Disease , Hot Temperature , Male , Motor Activity/physiology , Posterior Horn Cells/physiology , RNA, Messenger/metabolism , Random Allocation , Rats, Inbred WKY/psychology , Rats, Sprague-Dawley/psychology , Resilience, Psychological , Species Specificity , Swimming
5.
Curr Top Behav Neurosci ; 20: 251-80, 2014.
Article in English | MEDLINE | ID: mdl-24850075

ABSTRACT

The intensity and severity of perceived pain does not correlate consistently with the degree of peripheral or central nervous system tissue damage or with the intensity of primary afferent or spinal nociceptive neurone activity. In this respect, the modulation of pain by emotion and context is now widely recognized. In particular, stress, fear and anxiety exert potent, but complex, modulatory influences on pain. Stress can either suppress pain (stress-induced analgesia) or exacerbate it (stress-induced hyperalgesia; SIH) depending on the nature, duration and intensity of the stressor. Herein, we review the methods and models used to study the phenomenon of SIH in rodents and humans and then present a detailed discussion of our current understanding of neural substrates and neurobiological mechanisms. The review provides perspectives and challenges for the current and future treatment of pain and the co-morbidity of pain with stress-related psychiatric disorders including anxiety and depression.

6.
Eur J Neurosci ; 39(3): 435-43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24494683

ABSTRACT

The endogenous cannabinoid (endocannabinoid) system plays a key role in the modulation of aversive and nociceptive behaviour. The components of the endocannabinoid system are expressed throughout the hippocampus, a brain region implicated in both conditioned fear and pain. In light of evidence that pain can impact on the expression of fear-related behaviour, and vice versa, we hypothesised that exogenous administration of the endocannabinoid 2-arachidonoyl glycerol (2-AG) into the ventral hippocampus (vHip) would differentially regulate fear responding in the absence vs. the presence of formalin-evoked nociceptive tone. Fear-conditioned rats showed significantly increased freezing and a reduction in formalin-evoked nociceptive behaviour upon re-exposure to a context previously paired with footshock. Bilateral microinjection of 2-AG into the vHip significantly reduced contextually induced freezing in non-formalin-treated rats, and reduced formalin-evoked nociceptive behaviour in non-fear-conditioned rats. In contrast, 2-AG microinjection had no effect on fear responding in formalin-treated rats, and no effect on nociceptive behaviour in fear-conditioned rats. The inhibitory effect of 2-AG on fear-related behaviour, but not pain-related behaviour, was blocked by co-administration of the cannabinoid receptor 1 (CB1) antagonist/inverse agonist rimonabant. Tissue levels of the endocannabinoids N-arachidonoylethanolamide (anandamide, AEA) and 2-AG were similar in the vHip of fear-conditioned rats receiving formalin injection and the vHip of fear-conditioned rats receiving saline injection. However, the levels of AEA and 2-AG were significantly lower in the contralateral ventrolateral periaqueductal grey of formalin-treated fear-conditioned rats than in that of their saline-treated counterparts. These data suggest that 2-AG-CB1 receptor signalling in the vHip has an anti-aversive effect, and that this effect is abolished in the presence of a persistent pain state.


Subject(s)
Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Endocannabinoids/pharmacology , Fear/drug effects , Glycerides/pharmacology , Hippocampus/drug effects , Pain/physiopathology , Animals , Arachidonic Acids/administration & dosage , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Antagonists/pharmacology , Conditioning, Classical , Endocannabinoids/administration & dosage , Freezing Reaction, Cataleptic , Glycerides/administration & dosage , Hippocampus/physiopathology , Injections, Intraventricular , Mice , Nociception , Pain/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Pyrazoles/pharmacology , Rats , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant
7.
Pain ; 155(1): 69-79, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24076311

ABSTRACT

Pain is both a sensory and an emotional experience, and is subject to modulation by a number of factors including genetic background modulating stress/affect. The Wistar-Kyoto (WKY) rat exhibits a stress-hyper-responsive and depressive-like phenotype and increased sensitivity to noxious stimuli, compared with other rat strains. Here, we show that this genotype-dependent hyperalgesia is associated with impaired pain-related mobilisation of endocannabinoids and transcription of their synthesising enzymes in the rostral ventromedial medulla (RVM). Pharmacological blockade of the Cannabinoid1 (CB1) receptor potentiates the hyperalgesia in WKY rats, whereas inhibition of the endocannabinoid catabolising enzyme, fatty acid amide hydrolase, attenuates the hyperalgesia. The latter effect is mediated by CB1 receptors in the RVM. Together, these behavioural, neurochemical, and molecular data indicate that impaired endocannabinoid signalling in the RVM underpins hyper-responsivity to noxious stimuli in a genetic background prone to heightened stress/affect.


Subject(s)
Depression/psychology , Endocannabinoids/metabolism , Hyperkinesis/psychology , Medulla Oblongata/metabolism , Nociception/physiology , Signal Transduction/physiology , Animals , Cannabinoid Receptor Modulators/pharmacology , Disease Models, Animal , Endocannabinoids/genetics , Formaldehyde/toxicity , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , Medulla Oblongata/drug effects , Microdissection , Nociception/drug effects , Pain Measurement , Random Allocation , Rats , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects
8.
Pain ; 154(4): 576-585, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23414578

ABSTRACT

The basolateral amygdala (BLA) is a key substrate facilitating the expression of fear-conditioned analgesia (FCA). However, the neurochemical mechanisms in the BLA which mediate this potent suppression of pain responding during fear remain unknown. The present study investigated the role of cannabinoid1 (CB1) receptors and interactions with GABAergic (GABAA receptor) and glutamatergic (metabotropic glutamate receptor type 5; mGluR5) signalling in the BLA in formalin-evoked nociceptive behaviour and FCA in rats. Reexposure to a context previously paired with foot shock significantly reduced formalin-evoked nociceptive behaviour. Systemic or intra-BLA microinjection of the CB1 receptor antagonist/inverse agonist AM251 prevented this expression of FCA, while injection of AM251 into the central nucleus of the amygdala did not. The suppression of FCA by systemic AM251 administration was partially attenuated by intra-BLA administration of either the GABAA receptor antagonist bicuculline or the mGluR5 antagonist 2-methyl-6-(phenylethynyl) pyridine, (MPEP). Bilateral microinjection of MPEP, but not bicuculline, alone into the BLA enhanced formalin-evoked nociceptive behaviour. Postmortem analyses revealed that FCA was associated with a significant increase in tissue levels of anandamide in the BLA side contralateral to intraplantar formalin injection. In addition, fear-conditioned rats exhibited a robust formalin-induced increase in levels of 2-arachidonyl glycerol and N-palmitoylethanolamide in the ipsilateral and contralateral BLA, respectively. These data suggest that CB1 receptors in the BLA facilitate the expression of FCA, through a mechanism which is likely to involve the modulation of GABAergic and glutamatergic signalling.


Subject(s)
Amygdala/physiology , Analgesia , Conditioning, Psychological/physiology , Endocannabinoids/metabolism , Fear/physiology , Receptors, GABA/physiology , Receptors, Glutamate/physiology , Signal Transduction/physiology , Amygdala/drug effects , Animals , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Endocannabinoids/pharmacology , Ethanolamines/metabolism , Excitatory Amino Acid Agents/pharmacology , Fear/drug effects , Functional Laterality/drug effects , GABA Agents/pharmacology , Glycerides/pharmacology , Injections, Intraventricular , Male , Pain/psychology , Pain Measurement/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Receptor, Cannabinoid, CB1/physiology , Signal Transduction/drug effects
9.
J Neuroinflammation ; 9: 79, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22537429

ABSTRACT

BACKGROUND: Several factors contribute to the deterioration in synaptic plasticity which accompanies age and one of these is neuroinflammation. This is characterized by increased microglial activation associated with increased production of proinflammatory cytokines like interleukin-1ß (IL-1ß). In aged rats these neuroinflammatory changes are associated with a decreased ability of animals to sustain long-term potentiation (LTP) in the dentate gyrus. Importantly, treatment of aged rats with agents which possess anti-inflammatory properties to decrease microglial activation, improves LTP. It is known that endocannabinoids, such as anandamide (AEA), have anti-inflammatory properties and therefore have the potential to decrease the age-related microglial activation. However, endocannabinoids are extremely labile and are hydrolyzed quickly after production. Here we investigated the possibility that inhibiting the degradation of endocannabinoids with the fatty acid amide hydrolase (FAAH) inhibitor, URB597, could ameliorate age-related increases in microglial activation and the associated decrease in LTP. METHODS: Young and aged rats received subcutaneous injections of the FAAH inhibitor URB597 every second day and controls which received subcutaneous injections of 30% DMSO-saline every second day for 28 days. Long-term potentiation was recorded on day 28 and the animals were sacrificed. Brain tissue was analyzed for markers of microglial activation by PCR and for levels of endocannabinoids by liquid chromatography coupled to tandem mass spectrometry. RESULTS: The data indicate that expression of markers of microglial activation, MHCII, and CD68 mRNA, were increased in the hippocampus of aged, compared with young, rats and that these changes were associated with increased expression of the proinflammatory cytokines interleukin (IL)-1ß and tumor necrosis factor-α (TNFα) which were attenuated by treatment with URB597. Coupled with these changes, we observed an age-related decrease in LTP in the dentate gyrus which was partially restored in URB597-treated aged rats. The data suggest that enhancement of levels of endocannabinoids in the brain by URB597 has beneficial effects on synaptic function, perhaps by modulating microglial activation.


Subject(s)
Aging/drug effects , Amidohydrolases/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzamides/pharmacology , Carbamates/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Aging/pathology , Amidohydrolases/physiology , Animals , Hippocampus/enzymology , Hippocampus/pathology , Long-Term Potentiation/physiology , Male , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Rats , Rats, Wistar
10.
J Biol Chem ; 286(8): 6375-85, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21138836

ABSTRACT

Blockade of neurotransmitter release by botulinum neurotoxin type A (BoNT(A)) underlies the severe neuroparalytic symptoms of human botulism, which can last a few years. The structural basis for this remarkable persistence remains unclear. Herein, recombinant BoNT(A) was found to match the neurotoxicity of that from Clostridium botulinum, producing persistent cleavage of synaptosomal-associated protein of 25 kDa (SNAP-25) and neuromuscular paralysis. When two leucines near the C terminus of the protease light chain of A (LC(A)) were mutated, its inhibition of exocytosis was followed by fast recovery of intact SNAP-25 in cerebellar neurons and neuromuscular transmission in vivo. Deletion of 6-7 N terminus residues diminished BoNT(A) activity but did not alter the longevity of its SNAP-25 cleavage and neuromuscular paralysis. Furthermore, genetically fusing LC(E) to a BoNT(A) enzymically inactive mutant (BoTIM(A)) yielded a novel LC(E)-BoTIM(A) protein that targets neurons, and the BoTIM(A) moiety also delivers and stabilizes the inhibitory LC(E), giving a potent and persistent cleavage of SNAP-25 with associated neuromuscular paralysis. Moreover, its neurotropism was extended to sensory neurons normally insensitive to BoNT(E). LC(E-)BoTIM(A)(AA) with the above-identified dileucine mutated gave transient neuromuscular paralysis similar to BoNT(E), reaffirming that these residues are critical for the persistent action of LC(E)-BoTIM(A) as well as BoNT(A). LC(E)-BoTIM(A) inhibited release of calcitonin gene-related peptide from sensory neurons mediated by transient receptor potential vanilloid type 1 and attenuated capsaicin-evoked nociceptive behavior in rats, following intraplantar injection. Thus, a long acting, versatile composite toxin has been developed with therapeutic potential for pain and conditions caused by overactive cholinergic nerves.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Cerebellum/metabolism , Leucine , Neuromuscular Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Sensory Receptor Cells/metabolism , Synaptic Transmission/drug effects , Animals , Botulinum Toxins, Type A/genetics , Calcitonin/genetics , Calcitonin/metabolism , Cerebellum/cytology , Female , Male , Mice , Mutation , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Sensory Receptor Cells/cytology , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Time Factors
11.
Eur J Pain ; 14(5): 487-95, 2010 May.
Article in English | MEDLINE | ID: mdl-19786358

ABSTRACT

The basolateral amygdala (BLA) contains a high density of cannabinoid CB1 receptors and is critically involved in pain and fear-related behaviour. We investigated the effects of bilateral intra-BLA administration of the CB1 receptor antagonist/inverse agonist, rimonabant, on formalin-evoked nociceptive behaviour, fear-conditioned behaviour including analgesia, and associated brain regional alterations in Fos expression in rats. Intra-BLA administration of rimonabant significantly reduced formalin-evoked nociceptive behaviour in the absence, but not presence, of conditioned fear. Rimonabant attenuated a formalin-evoked reduction in freezing while emitting 22 kHz ultrasonic vocalisation in the early part of the fear expression trial. Formalin-evoked nociceptive behaviour was associated with increased Fos immunoreactivity (FI) in the CA2/3 region of the hippocampus and rostral ventromedial medulla, effects attenuated by intra-BLA rimonabant. Formalin also decreased FI in the cingulate cortex, an effect which was not observed in fear-conditioned rats. Contextually-induced fear was associated with increased FI in the dorsal caudal periaqueductal grey in the absence, but not presence, of formalin-evoked nociceptive tone. In conclusion, bilateral intra-BLA administration of rimonabant reduces nociceptive behaviour in a model of tonic, persistent inflammatory pain, an effect associated with reduced activation of neurons in the CA2/3 hippocampus and rostral ventromedial medulla. The data also provide evidence for differential pain- and fear-related brain regional activity in the presence or absence of contextually-induced aversion and nociceptive tone.


Subject(s)
Amygdala/drug effects , Behavior, Animal/drug effects , Fear/physiology , Neurons/drug effects , Pain Threshold/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Amygdala/metabolism , Analysis of Variance , Animals , Cannabinoid Receptor Antagonists , Catheters, Indwelling , Cell Count , Conditioning, Psychological/physiology , Formaldehyde , Hippocampus/drug effects , Hippocampus/metabolism , Immunohistochemistry , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Neurons/metabolism , Pain/chemically induced , Pain/metabolism , Pain Measurement , Periaqueductal Gray/drug effects , Periaqueductal Gray/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Receptors, Cannabinoid/metabolism , Rimonabant
SELECTION OF CITATIONS
SEARCH DETAIL
...